Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters










Publication year range
1.
PLoS One ; 4(6): e5963, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19536287

ABSTRACT

Mutations that cause a reduction in protein kinase A (PKA) activity have been shown to extend lifespan in yeast. Loss of function of mammalian RIIbeta, a regulatory subunit of PKA expressed in brain and adipose tissue, results in mice that are lean and insulin sensitive. It was therefore hypothesized that RIIB null (RIIbeta(-/-)) mice would express anti-aging phenotypes. We conducted lifespan studies using 40 mutant and 40 wild type (WT) littermates of equal gender numbers and found that both the median and maximum lifespans were significantly increased in mutant males compared to WT littermates. The median lifespan was increased from 884 days to 1005 days (p = 0.006 as determined by the log rank test) and the 80% lifespan (defined here as 80% deaths) was increased from 941 days to 1073 days (p = 0.004 as determined by the Wang-Allison test). There was no difference in either median or 80% lifespan in female genotypes. WT mice of both genders became increasingly obese with age, while mutant mice maintained their lean phenotype into old age. Adiposity was found to correlate with lifespan for males only. 50% of male mice between 30 and 35 g, corresponding to about 5% body fat, for either genotype lived over 1000 days. No male mouse outside of this weight range achieved this lifespan. During their last month of life, WT mice began losing weight (a total of 8% and 15% of body weight was lost for males and females, respectively), but RIIbeta(-/-) male mice maintained their lean body mass to end of life. This attenuation of decline was not seen in female mutant mice. Old male mutant mice were insulin sensitive throughout their life. Both genders showed modestly lower blood glucose levels in old mutants compared to WT. Male mutants were also resistant to age-induced fatty liver. Pathological assessment of tissues from end of life male mutant mice showed a decrease in tumor incidence, decreased severity of renal lesions, and a trend towards a decrease in age-related cardiac pathology. These findings help establish the highly conserved nature of PKA and suggest that disruption of PKA affects physiological mechanisms known to be associated with healthy aging.


Subject(s)
Aging , Cyclic AMP-Dependent Protein Kinases/physiology , Adipose Tissue/metabolism , Animals , Body Weight , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Genotype , Leptin/blood , Longevity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sex Factors
2.
Diabetes ; 50(11): 2555-62, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11679434

ABSTRACT

The mechanisms by which obesity contributes to diabetic phenotypes remain unclear. We evaluated the role of protein kinase A (PKA) signaling events in mediating diabetes associated with obesity. PKA comprises two regulatory subunits and two catalytic subunits and is activated by cAMP. The RIIbeta regulatory subunit is abundantly expressed in adipose tissue and brain. Knockout mice lacking this subunit are lean and display remarkable resistance to diet-induced obesity. We investigated whether these mice were also resistant to diet-induced diabetes and whether this effect was dependent on reduced adiposity. Mice were fed a high-fat, high-carbohydrate diet and weight gain and diabetes phenotypes were examined. RIIbeta(-/-) mice displayed decreased body weights, reduced insulin levels, improved insulin sensitivity, and improved total-body glucose disposal as compared with wild-type controls. Plasma levels of VLDL and LDL cholesterol were also reduced in high fat-fed RIIbeta(-/-) mice compared with wild-type mice. Taken together, these data demonstrate that loss of RIIbeta protects mice from diet-induced obesity, insulin resistance, and dyslipidemia.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Diet/adverse effects , Hyperlipidemias/etiology , Hyperlipidemias/prevention & control , Insulin Resistance/physiology , Mutation/physiology , Adipose Tissue/anatomy & histology , Animals , Body Weight , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Diabetes Mellitus/etiology , Diabetes Mellitus/genetics , Female , Glucose/metabolism , Insulin/pharmacology , Lipids/blood , Longitudinal Studies , Male , Mice , Mice, Knockout/genetics , Phenotype , Reference Values
3.
Nat Genet ; 25(4): 448-52, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10932193

ABSTRACT

Ca2+/calmodulin-dependent protein kinase IV (Camk4; also known as CaMKIV), a multifunctional serine/threonine protein kinase with limited tissue distribution, has been implicated in transcriptional regulation in lymphocytes, neurons and male germ cells. In the mouse testis, however, Camk4 is expressed in spermatids and associated with chromatin and nuclear matrix. Elongating spermatids are not transcriptionally active, raising the possibility that Camk4 has a novel function in male germ cells. To investigate the role of Camk4 in spermatogenesis, we have generated mice with a targeted deletion of the gene Camk4. Male Camk4-/- mice are infertile with impairment of spermiogenesis in late elongating spermatids. The sequential deposition of sperm basic nuclear proteins on chromatin is disrupted, with a specific loss of protamine-2 and prolonged retention of transition protein-2 (Tnp2) in step-15 spermatids. Protamine-2 is phosphorylated by Camk4 in vitro, implicating a connection between Camk4 signalling and the exchange of basic nuclear proteins in mammalian male germ cells. Defects in protamine-2 have been identified in sperm of infertile men, suggesting that our results may have clinical implications for the understanding of human male infertility.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/genetics , Nuclear Proteins/metabolism , Spermatogenesis/physiology , Spermatozoa/metabolism , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Molecular Sequence Data , Phosphorylation , Protamines/genetics , Protamines/metabolism , Sperm Count , Spermatozoa/cytology , Spermatozoa/enzymology , Testis/growth & development , Testis/metabolism
4.
Proc Natl Acad Sci U S A ; 97(12): 6433-8, 2000 Jun 06.
Article in English | MEDLINE | ID: mdl-10841548

ABSTRACT

The catalytic subunits of protein kinase A are transcribed in all mouse tissues from two distinct genes that code for the Calpha and Cbeta isoforms. Alternative promoters exist for the Cbeta gene that are used in a tissue-specific fashion and give rise to variants that differ in their amino-terminal sequences. We have characterized an alternative promoter that is present in the first intron of the Calpha gene and is transcriptionally active in male germ cells. Transcription from this promoter is coincident with the appearance of pachytene spermatocytes and leads to a Calpha protein (Calpha2) that contains a distinctive 7 amino acid amino-terminus differing from the 14 amino acid amino-terminus of Calpha1. The Calpha2 protein does not contain the myristylation signal present on Calpha1 and migrates at a lower molecular weight on SDS/PAGE gels. By Western blotting, we estimate that most or all of the Calpha protein present in mature sperm is Calpha2. The amino-terminal sequence of Calpha2 is similar to that of ovine sperm C as previously reported [San Agustin, J. T., Leszyk, J. D., Nuwaysir, L. M. & Witman, G. B. (1998) J. Biol. Chem. 273, 24874-24883], and we show by cDNA cloning that human sperm also express a highly related Calpha2 homolog. The Calpha2 subunit forms holoenzymes with either RIIalpha or RIalpha, and both activate at the same concentration of cyclic nucleotide. Because protein kinase A is thought to play a pivotal role in sperm motility and capacitation, the distinctive biochemical properties of the unmyristylated Calpha2 may be essential for fertility in the male.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Isoenzymes/genetics , Spermatozoa/enzymology , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Catalytic Domain , Chromosome Mapping , Conserved Sequence , Enzyme Activation , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Spermatozoa/physiology , Testis/enzymology , Transcription, Genetic
5.
J Neurosci ; 20(10): RC75, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10783399

ABSTRACT

Both in vitro and in vivo evidence indicate that cAMP-dependent protein kinase (PKA) mediates some of the acute and chronic cellular responses to alcohol. However, it is unclear whether PKA regulates voluntary alcohol consumption. We therefore studied alcohol consumption by mice that completely lack the regulatory IIbeta (RIIbeta) subunit of PKA as a result of targeted gene disruption. Here we report that RIIbeta knockout mice (RIIbeta-/-) showed incr eased consumption of solutions containing 6, 10, and 20% (v/v) ethanol when compared with wild-type mice (RIIbeta+/+). On the other hand, RIIbeta-/- mice showed normal consumption of solutions containing either sucrose or quinine. When compared with wild-type mice, the RIIbeta-/- mice were found to be less sensitive to the sedative effects of ethanol as measured by more rapid recovery from ethanol-induced sleep, even though plasma ethanol concentrations did not differ significantly from those of controls. Finally, both RIbeta- and catylatic subunit beta1-deficient mice showed normal voluntary consumption of ethanol, indicating that increased ethanol consumption is not a general characteristic associated with deletion of PKA subunits. These data demonstrate a role for the RIIbeta subunit of PKA in regulating voluntary consumption of alcohol and sensitivity to the intoxication effects that are produced by this drug.


Subject(s)
Alcohol Drinking/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Ethanol/pharmacology , Hypnotics and Sedatives/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Cyclic AMP-Dependent Protein Kinases/metabolism , Ethanol/metabolism , Hypnotics and Sedatives/metabolism , Mice , Mice, Knockout , Quinine/pharmacology , Sucrose/pharmacology
6.
Mol Cell Biol ; 20(10): 3442-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10779334

ABSTRACT

Protein kinase inhibitor (PKI) is a potent endogenous inhibitor of the cyclic AMP (cAMP)-dependent protein kinase (PKA). It functions by binding the free catalytic (C) subunit with a high affinity and is also known to export nuclear C subunit to the cytoplasm. The significance of these actions with respect to PKI's physiological role is not well understood. To address this, we have generated by homologous recombination mutant mice that are deficient in PKIalpha, one of the three isoforms of PKI. The mice completely lack PKI activity in skeletal muscle and, surprisingly, show decreased basal and isoproterenol-induced gene expression in muscle. Further examination revealed reduced levels of the phosphorylated (active) form of the transcription factor CREB (cAMP response element binding protein) in the knockouts. This phenomenon stems, at least in part, from lower basal PKA activity levels in the mutants, arising from a compensatory increase in the level of the RIalpha subunit of PKA. The deficit in gene induction, however, is not easily explained by current models of PKI function and suggests that PKI may play an as yet undescribed role in PKA signaling.


Subject(s)
Carrier Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins , Muscle, Skeletal/physiology , Animals , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Inhibitors , Gene Expression Regulation , Homozygote , Isoproterenol/pharmacology , Mice , Mice, Knockout , Phosphorylation , Protein Isoforms/genetics , Signal Transduction , Transcriptional Activation
7.
J Biol Chem ; 274(51): 36281-7, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593917

ABSTRACT

Targeted disruption of the RIIbeta subunit of protein kinase A (PKA) produces lean mice that resist diet-induced obesity. In this report we examine the effects of the RIIbeta knockout on white adipose tissue physiology. Loss of RIIbeta is compensated by an increase in the RIalpha isoform, generating an isoform switch from a type II to a type I PKA. Type I holoenzyme binds cAMP more avidly and is more easily activated than the type II enzyme. These alterations are associated with increases in both basal kinase activity and the basal rate of lipolysis, possibly contributing to the lean phenotype. However, the ability of both beta(3)-selective and nonspecific beta-adrenergic agonists to stimulate lipolysis is markedly compromised in mutant white adipose tissue. This defect was found in vitro and in vivo and does not result from reduced expression of beta-adrenergic receptor or hormone-sensitive lipase genes. In contrast, beta-adrenergic stimulated gene transcription remains intact, and the expression of key genes involved in lipid metabolism is normal under both fasted and fed conditions. We suggest that the R subunit isoform switch disrupts the subcellular localization of PKA that is required for efficient transduction of signals that modulate lipolysis but not for those that mediate gene expression.


Subject(s)
Adipose Tissue/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Lipolysis/genetics , Obesity/genetics , Obesity/metabolism , Animals , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Cyclic AMP-Dependent Protein Kinase Type II , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation
8.
J Biol Chem ; 274(34): 24131-6, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10446185

ABSTRACT

Cyclic AMP stimulates sperm motility in a variety of mammalian species, but the molecular details of the intracellular signaling pathway responsible for this effect are unclear. The type IIalpha isoform of protein kinase A (PKA) is induced late in spermatogenesis and is thought to localize PKA to the flagellar apparatus where it binds cAMP and stimulates motility. A targeted disruption of the type IIalpha regulatory subunit (RIIalpha) gene allowed us to examine the role of PKA localization in sperm motility and fertility. In wild type sperm, PKA is found primarily in the detergent-resistant particulate fraction and localizes to the mitochondrial-containing midpiece and the principal piece. In mutant sperm, there is a compensatory increase in RIalpha protein and a dramatic relocalization of PKA such that the majority of the holoenzyme now appears in the soluble fraction and colocalizes with the cytoplasmic droplet. Unexpectedly the RIIalpha mutant mice are fertile and have no significant changes in sperm motility. Our results demonstrate that the highly localized pattern of PKA seen in mature sperm is not essential for motility or fertilization.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Fertility , Sperm Motility , Spermatozoa/enzymology , Acrosome/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/physiology , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mutation , Structure-Activity Relationship
9.
J Biol Chem ; 274(10): 6381-7, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10037729

ABSTRACT

Regulation of protein kinase A by subcellular localization may be critical to target catalytic subunits to specific substrates. We employed epitope-tagged catalytic subunit to correlate subcellular localization and gene-inducing activity in the presence of regulatory subunit or protein kinase inhibitor (PKI). Transiently expressed catalytic subunit distributed throughout the cell and induced gene expression. Co-expression of regulatory subunit or PKI blocked gene induction and prevented nuclear accumulation. A mutant PKI lacking the nuclear export signal blocked gene induction but not nuclear accumulation, demonstrating that nuclear export is not essential to inhibit gene induction. When the catalytic subunit was targeted to the nucleus with a nuclear localization signal, it was not sequestered in the cytoplasm by regulatory subunit, although its activity was completely inhibited. PKI redistributed the nuclear catalytic subunit to the cytoplasm and blocked gene induction, demonstrating that the nuclear export signal of PKI can override a strong nuclear localization signal. With increasing PKI, the export process appeared to saturate, resulting in the return of catalytic subunit to the nucleus. These results demonstrate that both the regulatory subunit and PKI are able to completely inhibit the gene-inducing activity of the catalytic subunit even when the catalytic subunit is forced to concentrate in the nuclear compartment.


Subject(s)
Carrier Proteins , Cell Nucleus/enzymology , Cyclic AMP-Dependent Protein Kinases/analysis , Intracellular Signaling Peptides and Proteins , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation , Promoter Regions, Genetic
10.
Recent Prog Horm Res ; 53: 139-59; discussion 160-1, 1998.
Article in English | MEDLINE | ID: mdl-9769707

ABSTRACT

The major regulator of lipolysis in white adipocytes and brown adipocytes is cAMP and the actions of cAMP are mediated by protein kinase A (PKA). Multiple subunits of PKA, including RII beta, R1 alpha, C alpha, and C beta 1, are expressed in fat cells but the major holoenzyme assembled under normal conditions contains RII beta and C alpha. Targeted disruption of the RII beta gene in mice revealed that both white and brown adipocytes are capable of compensating by increasing the level of RI alpha. Nevertheless, the mice display a lean phenotype, have an elevated metabolic rate due to activation and induction of uncoupling protein in brown fat, and are resistant to diet-induced obesity and insulin resistance. Although the metabolic disturbances in white and brown fat tissue may explain most of the phenotypic changes, the loss of neuronal expression of RII beta may also contribute to the alterations in energy balance. Specific neuronal defects have been characterized that prevent the normal changes in gene expression seen with drugs that act through the dopaminergic pathway. The RII beta mutant mouse provides an interesting model of obesity resistance and demonstrates that chronic changes in the PKA signaling system can lead to stable alterations in energy storage and utilization.


Subject(s)
Adipose Tissue/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/physiology , Animals , Mice
11.
J Neurosci ; 18(10): 3639-49, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9570795

ABSTRACT

Motor behavior is modulated by dopamine-responsive neurons in the striatum, where dopaminergic signaling uses G-protein-coupled pathways, including those that result in the activation of cAMP-dependent protein kinase (PKA). The RIIbeta isoform of PKA is highly enriched in the striatum, and targeted disruption of the RIIbeta gene in mice leads to a dramatic reduction in total PKA activity in this region. Although the mutant mice show typical locomotor responses after acute administration of dopaminergic drugs, they display abnormalities in two experience-dependent locomotor behaviors: training on the rotarod task and locomotor sensitization to amphetamine. In addition, amphetamine induction of fos is absent, and the basal expression of dynorphin mRNA is reduced in the striatum. These results demonstrate that motor learning and the regulation of neuronal gene expression require RIIbeta PKA, whereas the acute locomotor effects of dopaminergic drugs are relatively unaffected by this PKA deficiency.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Motor Activity/physiology , Amphetamine/pharmacology , Animals , Behavior, Animal/physiology , Corpus Striatum/cytology , Corpus Striatum/drug effects , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/physiology , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Dynorphins/genetics , Fertility/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , In Situ Hybridization , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Locomotion/drug effects , Locomotion/physiology , Longevity/genetics , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Neurons/enzymology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism
12.
Hum Mol Genet ; 7(6): 959-67, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9580659

ABSTRACT

X-linked spinal and bulbar muscular atrophy (SBMA) is caused by a CAG repeat expansion in the first exon of the androgen receptor (AR) gene. Disease-associated alleles (37-66 CAGs) change in length when transmitted from parents to offspring, with a significantly greater tendency to shift size when inherited paternally. As transgenic mice carrying human AR cDNAs with 45 and 66 CAG repeats do not display repeat instability, we attempted to model trinucleotide repeat instability by generating transgenic mice with yeast artificial chromosomes (YACs) carrying AR CAG repeat expansions in their genomic context. Studies of independent lines of AR YAC transgenic mice with CAG 45 alleles reveal intergenerational instability at an overall rate of approximately 10%. We also find that the 45 CAG repeat tracts are significantly more unstable with maternal transmission and as the transmitting mother ages. Of all the CAG/CTG repeat transgenic mice produced to date the AR YAC CAG 45 mice are unstable with the smallest trinucleotide repeat mutations, suggesting that the length threshold for repeat instability in the mouse may be lowered by including the appropriate flanking human DNA sequences. By sequence-tagged site content analysis and long range mapping we determined that one unstable transgenic line has integrated an approximately 70 kb segment of the AR locus due to fragmentation of the AR YAC. Identification of the cis -acting elements that permit CAG tract instability and the trans -acting factors that modulate repeat instability in the AR YAC CAG 45 mice may provide insights into the molecular basis of trinucleotide repeat instability in humans.


Subject(s)
Muscular Atrophy, Spinal/genetics , Receptors, Androgen/genetics , Trinucleotide Repeats , Age Factors , Alleles , Animals , Chromosome Mapping , Chromosomes, Artificial, Yeast , Disease Models, Animal , Female , Gene Dosage , Gene Expression , Humans , Mice , Mice, Transgenic , Mosaicism/genetics , Sequence Tagged Sites , Sex Factors , X Chromosome
13.
J Neurosci ; 18(6): 2108-17, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9482797

ABSTRACT

Developing sensory systems are sculpted by an activity-dependent strengthening and weakening of connections. Long-term potentiation (LTP) and depression (LTD) in vitro have been proposed to model this experience-dependent circuit refinement. We directly compared LTP and LTD induction in vitro with plasticity in vivo in the developing visual cortex of a mouse mutant of protein kinase A (PKA), a key enzyme implicated in the plasticity of a diverse array of systems. In mice lacking the RIbeta regulatory subunit of PKA, we observed three abnormalities of synaptic plasticity in layer II/III of visual cortex in vitro. These included an absence of (1) extracellularly recorded LTP, (2) depotentiation or LTD, and (3) paired-pulse facilitation. Potentiation was induced, however, by pairing low-frequency stimulation with direct depolarization of individual mutant pyramidal cells. Together these findings suggest that the LTP defect in slices lacking PKA RIbeta lies in the transmission of sufficient net excitation through the cortical circuit. Nonetheless, functional development and plasticity of visual cortical responses in vivo after monocular deprivation did not differ from normal. Moreover, the loss of all responsiveness to stimulation of the originally deprived eye in most cortical cells could be restored by reverse suture of eyelids during the critical period in both wild-type and mutant mice. Such an activity-dependent increase in response would seem to require a mechanism like potentiation in vivo. Thus, the RIbeta isoform of PKA is not essential for ocular dominance plasticity, which can proceed despite defects in several common in vitro models of neural plasticity.


Subject(s)
Aging/physiology , Cyclic AMP-Dependent Protein Kinases/deficiency , Neuronal Plasticity/physiology , Visual Cortex/physiology , Action Potentials/physiology , Animals , Cyclic AMP-Dependent Protein Kinase RIbeta Subunit , Extracellular Space/physiology , Functional Laterality/physiology , Long-Term Potentiation/physiology , Mice , Ocular Physiological Phenomena , Patch-Clamp Techniques , Reference Values , Sensory Deprivation/physiology , Synapses/physiology , Vision, Monocular/physiology , Visual Cortex/growth & development
14.
Proc Natl Acad Sci U S A ; 94(24): 13311-6, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9371842

ABSTRACT

Muscarinic acetylcholine receptors are members of the G protein-coupled receptor superfamily expressed in neurons, cardiomyocytes, smooth muscle, and a variety of epithelia. Five subtypes of muscarinic acetylcholine receptors have been discovered by molecular cloning, but their pharmacological similarities and frequent colocalization make it difficult to assign functional roles for individual subtypes in specific neuronal responses. We have used gene targeting by homologous recombination in embryonic stem cells to produce mice lacking the m1 receptor. These mice show no obvious behavioral or histological defects, and the m2, m3, and m4 receptors continue to be expressed in brain with no evidence of compensatory induction. However, the robust suppression of the M-current potassium channel activity evoked by muscarinic agonists in sympathetic ganglion neurons is completely lost in m1 mutant mice. In addition, both homozygous and heterozygous mutant mice are highly resistant to the seizures produced by systemic administration of the muscarinic agonist pilocarpine. Thus, the m1 receptor subtype mediates M current modulation in sympathetic neurons and induction of seizure activity in the pilocarpine model of epilepsy.


Subject(s)
Receptors, Muscarinic/physiology , Seizures/genetics , Animals , Behavior, Animal/drug effects , Disease Susceptibility , Kainic Acid/pharmacology , Ligands , Mice , Mice, Knockout , Muscarinic Antagonists/pharmacology , Pilocarpine/pharmacology , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism
15.
J Biol Chem ; 272(47): 29560-5, 1997 Nov 21.
Article in English | MEDLINE | ID: mdl-9368018

ABSTRACT

We have previously characterized two murine cAMP-dependent protein kinase catalytic subunit genes, Calpha and Cbeta1. Targeted disruption of the Cbeta1 promoter revealed two splice variants of the Cbeta catalytic subunit gene (designated Cbeta2 and Cbeta3) that continue to be expressed. These variants arise from unique promoters and are brain-specific. Cbeta2 is expressed in several discrete areas in the limbic system. These include the lateral septum, the bed nucleus of the stria terminalis, the ventral medial hypothalamus, and the amygdala. In the neocortex, expression is highest in cortical areas such as the prefrontal and insular cortex that are associated limbic structures. By contrast, Cbeta1 is most highly expressed in the cortex and hippocampus and is also present in all non-neuronal tissues examined. Cbeta3 is expressed at very low levels with wide distribution throughout the brain. Both the Cbeta2 and Cbeta3 variants are enzymatically active and induce gene expression in transient transfections with a cAMP response element-reporter construct. This activity is inhibited by protein kinase A regulatory subunits, the protein kinase inhibitor, and the chemical inhibitor H-89. We also demonstrate that Cbeta1 is myristoylated at the amino terminus like the Calpha isoform, but neither Cbeta2 nor Cbeta3 is myristoylated. The discrete expression of Cbeta variants in the brain suggests specific functional roles in neuronal signaling.


Subject(s)
Alternative Splicing , Brain/enzymology , Cyclic AMP-Dependent Protein Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Chromosome Mapping , Gene Library , In Situ Hybridization , Mice , Molecular Sequence Data , Myristic Acid/metabolism
16.
Proc Natl Acad Sci U S A ; 94(22): 12157-61, 1997 Oct 28.
Article in English | MEDLINE | ID: mdl-9342379

ABSTRACT

The antipsychotic drug, haloperidol, elicits the expression of neurotensin and c-fos mRNA in the dorsal lateral region of the striatum and produces an acute cataleptic response in rodents that correlates with the motor side effects of haloperidol in humans. Mice harboring a targeted disruption of the RIIbeta subunit of protein kinase A have a profound deficit in cAMP-stimulated kinase activity in the striatum. When treated with haloperidol, RIIbeta mutant mice fail to induce either c-fos or neurotensin mRNA and the acute cataleptic response is blocked. However, both wild-type and mutant mice become cataleptic when neurotensin peptide is directly injected into the lateral ventricle, demonstrating that the kinase deficiency does not interfere with the action of neurotensin but rather its synthesis and release. These results establish a direct role for protein kinase A as a mediator of haloperidol induced gene induction and cataleptic behavior.


Subject(s)
Antipsychotic Agents/pharmacology , Catalepsy , Corpus Striatum/drug effects , Cyclic AMP-Dependent Protein Kinases/deficiency , Gene Expression Regulation/drug effects , Haloperidol/pharmacology , Animals , Behavior, Animal/drug effects , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Cyclic AMP-Dependent Protein Kinases/genetics , Mice , Mice, Mutant Strains , Neurotensin/biosynthesis , Neurotensin/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/isolation & purification , Receptors, Dopamine D2/metabolism , Signal Transduction , Sulpiride/metabolism , Transcription, Genetic , Transcriptional Activation
17.
Proc Natl Acad Sci U S A ; 94(20): 11067-72, 1997 Sep 30.
Article in English | MEDLINE | ID: mdl-9380760

ABSTRACT

Preferential phosphorylation of specific proteins by cAMP-dependent protein kinase (PKA) may be mediated in part by the anchoring of PKA to a family of A-kinase anchor proteins (AKAPs) positioned in close proximity to target proteins. This interaction is thought to depend on binding of the type II regulatory (RII) subunits to AKAPs and is essential for PKA-dependent modulation of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/kainate receptor, the L-type Ca2+ channel, and the KCa channel. We hypothesized that the targeted disruption of the gene for the ubiquitously expressed RIIalpha subunit would reveal those tissues and signaling events that require anchored PKA. RIIalpha knockout mice appear normal and healthy. In adult skeletal muscle, RIalpha protein levels increased to partially compensate for the loss of RIIalpha. Nonetheless, a reduction in both catalytic (C) subunit protein levels and total kinase activity was observed. Surprisingly, the anchored PKA-dependent potentiation of the L-type Ca2+ channel in RIIalpha knockout skeletal muscle was unchanged compared with wild type although it was more sensitive to inhibitors of PKA-AKAP interactions. The C subunit colocalized with the L-type Ca2+ channel in transverse tubules in wild-type skeletal muscle and retained this localization in knockout muscle. The RIalpha subunit was shown to bind AKAPs, although with a 500-fold lower affinity than the RIIalpha subunit. The potentiation of the L-type Ca2+ channel in RIIalpha knockout mouse skeletal muscle suggests that, despite a lower affinity for AKAP binding, RIalpha is capable of physiologically relevant anchoring interactions.


Subject(s)
Calcium Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Muscle, Skeletal/enzymology , Animals , Calcium Channels/physiology , Calcium Channels, L-Type , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Ion Channel Gating , Mice , Mice, Knockout , Muscle, Skeletal/physiology
18.
J Neurosci ; 17(19): 7462-70, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9295392

ABSTRACT

To assess the contribution of PKA to injury-induced inflammation and pain, we evaluated nociceptive responses in mice that carry a null mutation in the gene that encodes the neuronal-specific isoform of the type I regulatory subunit (RIbeta) of PKA. Acute pain indices did not differ in the RIbeta PKA mutant mice compared with wild-type controls. However, tissue injury-evoked persistent pain behavior, inflammation of the hindpaw, and ipsilateral dorsal horn Fos immunoreactivity was significantly reduced in the mutant mice, as was plasma extravasation induced by intradermal injection of capsaicin into the paw. The enhanced thermal sensitivity observed in wild-type mice after intraplantar or intrathecal (spinal) administration of prostaglandin E2 was also reduced in mutant mice. In contrast, indices of pain behavior produced by nerve injury were not altered in the mutant mice. Thus, RIbeta PKA is necessary for the full expression of tissue injury-evoked (nociceptive) pain but is not required for nerve injury-evoked (neuropathic) pain. Because the RIbeta subunit is only present in the nervous system, including small diameter trkA receptor-positive dorsal root ganglion cells, we suggest that in inflammatory conditions, RIbeta PKA is specifically required for nociceptive processing in the terminals of small-diameter primary afferent fibers.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Isoenzymes/genetics , Mutation , Nociceptors/physiology , Sciatic Nerve/injuries , Wounds, Nonpenetrating/pathology , Animals , Capsaicin/pharmacology , Cyclic AMP-Dependent Protein Kinase RIbeta Subunit , Cyclic AMP-Dependent Protein Kinases/metabolism , Differential Threshold , Dinoprostone/pharmacology , Gene Expression , Gene Targeting , Hot Temperature , Mice , Mice, Mutant Strains , Neuritis/etiology , Neuritis/physiopathology , Pain , Sensory Thresholds/drug effects , Transgenes/genetics , Wounds, Nonpenetrating/physiopathology
19.
Curr Opin Neurobiol ; 7(3): 397-403, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9232801

ABSTRACT

In mammals, the cAMP-dependent protein kinase (PKA) family of enzymes is assembled from the products of four regulatory and two catalytic subunit genes, all of which are expressed in neurons. Specific isoforms of PKA display differences in biochemical properties and subcellular localization, but it has been difficult to ascribe specific physiological functions to any given isoform. The recent development of gene knockout and transgenic mouse models has allowed for a more integrated examination of the in vivo roles of specific PKA isoforms in gene expression, synaptic plasticity, and behaviour.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Neural Pathways/physiology , Animals , Hippocampus/metabolism , Mice , Models, Biological
20.
J Biol Chem ; 272(7): 3993-8, 1997 Feb 14.
Article in English | MEDLINE | ID: mdl-9020105

ABSTRACT

The cAMP-dependent protein kinase holoenzyme is assembled from regulatory (R) and catalytic (C) subunits that are expressed in tissue-specific patterns. Despite the dispersion of the R and C subunit genes to different chromosomal loci, mechanisms exist that coordinately regulate the intracellular levels of R and C protein such that cAMP-dependent regulation is preserved. We have created null mutations in the RIbeta and RIIbeta regulatory subunit genes in mice, and find that both result in an increase in the level of RIalpha protein in tissues that normally express the beta isoforms. Examination of RIalpha mRNA levels and the rates of RIalpha protein synthesis in wild type and RIIbeta mutant mice reveals that the mechanism of this biochemical compensation by RIalpha does not involve transcriptional or translational control. These in vivo findings are consistent with observations made in cell culture, where we demonstrate that the overexpression of Calpha in NIH 3T3 cells results in increased RIalpha protein without increases in the rate of RIalpha synthesis or the level of RIalpha mRNA. Pulse-chase experiments reveal a 4-5-fold increase in the half-life of RIalpha protein as it becomes incorporated into the holoenzyme. Compensation by RIalpha stabilization may represent an important biological mechanism that safeguards cells from unregulated catalytic subunit activity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , 3T3 Cells , Adipose Tissue/enzymology , Animals , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/genetics , Mice , Mice, Mutant Strains , Protein Biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...